Open Access

A random forest classifier predicts recurrence risk in patients with ovarian cancer

  • Authors:
    • Li Cheng
    • Lin Li
    • Liling Wang
    • Xiaofang Li
    • Hui Xing
    • Jinting Zhou
  • View Affiliations

  • Published online on: July 19, 2018     https://doi.org/10.3892/mmr.2018.9300
  • Pages: 3289-3297
  • Copyright: © Cheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer (OC) is associated with a poor prognosis due to difficulties in early detection. The aims of the present study were to construct a recurrence risk prediction model and to reveal important OC genes or pathways. RNA sequencing data was obtained for 307 OC samples, and the corresponding clinical data were downloaded from The Cancer Genome Atlas database. Additionally, two validation datasets, GSE44104 (20 recurrent and 40 non‑recurrent OC samples) and GSE49997 (204 OC samples), were obtained from the Gene Expression Omnibus database. Differentially expressed genes were screened using the differential expression via distance synthesis algorithm, followed by gene ontology enrichment analysis and weighted gene coexpression network analysis (WGCNA). Furthermore, subnetwork analysis was conducted for the protein‑protein interaction (PPI) network using the BioNet package. Finally, a random forest classifier was constructed based on the subnetwork nodes, and its reliability was validated using the GSE44104 and GSE49997 validation datasets. A total of 44 upregulated and 117 downregulated genes were identified in the recurrent samples. Enrichment analysis indicated that cytochrome P450 family 17 subfamily A member 1 (CYP17A1) was associated with ‘positive regulation of steroid hormone biosynthetic processes’. WGCNA identified turquoise and grey modules that were significantly correlated with status and prognosis. A significant PPI subnetwork containing 16 nodes was also identified, including: Transcription factor GATA‑4; fibroblast growth factor 9; aromatase; 3β‑hydroxysteroid dehydrogenase/δ5‑4‑isomerase type 2; corticosteroid 11β‑dehydrogenase isozyme 1; CYP17A1; pituitary homeobox 2; left‑right determination factor 1; homeobox protein ARX; estrogen receptor β; steroidogenic factor 1; forkhead box protein L2; myocardin; steroidogenic acute regulatory protein mitochondrial; vesicular inhibitory amino acid transporter; and twist‑related protein 1. A random forest classifier was constructed using the subnetwork nodes as feature genes, which exhibited a 92% true positive rate when classifying recurrent and non‑recurrent OC samples. The classifying efficiency of the random forest classifier was validated using the two other independent datasets. Overall, 44 upregulated and 117 downregulated genes associated with OC recurrence were identified. Furthermore, the 16 subnetwork node genes that were identified may be important molecules in OC recurrence.

Introduction

Ovarian cancer (OC), which frequently occurs in postmenopausal women (1), is a cancer with no apparent symptoms until it reaches an advanced stage. The symptoms include bloating, abdominal swelling, pelvic pain and loss of appetite (2). The three most common OC subtypes are high-grade serous carcinomas, sex cord stromal tumors and germ cell tumors (3), which may metastasize to the peritoneum, liver, lungs or lymph nodes (4). OC is difficult to detect and metastasizes early in disease progression; therefore, patients with OC frequently have a poor prognosis (5). Globally, OC affects 1.2 million women and led to 161,100 mortalities in 2015 (6,7). Thus, obtaining an improved understanding of OC progression and recurrence is of great importance for improving its prognosis.

Previously, genes affecting OC were identified, including p21-activated kinase 4 (Pak4), cyclin E1 (CCNE1), RNA binding motif protein 3 (RBM3), YY1 associated protein 1 (YAP) and prominin-1 (CD133). Pak4 overexpression has been reported to contribute to OC cell migration, invasion and proliferation, thus making it a promising prognostic indicator and therapeutic target (8). CCNE1 amplification has been demonstrated to markedly reduce disease-free survival and overall survival, thus indicating that CCNE1-targeted treatment may benefit patients with OC who have upregulated CCNE1 expression (9,10). In epithelial ovarian cancer, RBM3 expression has been associated with cisplatin sensitivity and correlated with a positive patient prognosis (11). Furthermore, YAP has been associated with cell growth and tumorigenesis, and its coexpression with TEA domain transcription factor 4 serves as a predictor of a poor outcome (12,13). CD133 also serves as a predictor of poor OC patient survival, thus suggesting that it may serve as a biomarker of cancer stem cells during disease (14). However, the further identification of prognostic indicators and their potential uses is required.

In recent years, bioinformatics analysis of expression profile data has been gradually used to examine the pathogenesis of human diseases (15). It is known that the progress of a disease is usually mediated by multiple relevant genes and not by a single gene (16,17). Therefore, the present study was designed to mine subnetwork features and build a model to assess OC recurrence risk. In the present study, OC expression profiles were downloaded from a public database, and differentially expressed genes (DEGs) were analyzed and functionally enriched. Following identification of a functional subnetwork, a random forest classifier was constructed and validated. It was considered that this constructed classifier may provide an improved approach for predicting the prognoses of patients with OC.

Materials and methods

Data source

RNA sequence data from 307 OC samples and their corresponding clinical data (including patient vital status and overall survival) were downloaded from The Cancer Genome Atlas (TCGA; http://cancergenome.nih.gov) database. Based on patient vital status, 180 recurrent samples and 72 disease-free samples were identified. Additionally, the GSE44104 dataset [including 20 recurrent and 40 non-recurrent OC samples; platform, (HG-U133_Plus_2) Affymetrix Human Genome U133 Plus 2.0 Array, Affymetrix; Thermo Fisher Scientific, Inc., Waltham, MA, USA] and the GSE49997 dataset (including 204 OC samples; platform, GPL2986 ABI Human Genome Survey Microarray version 2, Applied Biosystems; Thermo Fisher Scientific, Inc.) were obtained from the Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo) database and utilized as validation sets.

Data preprocessing and DEG screening

Using a z-score algorithm (18), the expression value of each gene was normalized to a normal distribution (mean=0; variance=1). The samples were subsequently analyzed using the differential expression via distance synthesis (DEDS) algorithm, which may be applied to obtain differential expression levels via the distance synthesis of relevant data (19). This approach was used to screen DEGs in the recurrent samples relative to the disease-free samples.

Functional enrichment analysis

Gene ontology (GO; http://www.geneontology.org) analysis may be used to predict the potential functions of gene products (20). Using GO Term Finder (http://search.cpan.org/dist/GO-TermFinder/) as previously described (21), upregulated and downregulated genes were separately enriched. Functional terms with P<0.05 and an association with at least three genes were selected as significant terms.

Weighted gene coexpression network analysis (WGCNA)

Genes may jointly influence alterations in functional terms through their interactions, with functional consistencies between genes also confirmed by significant expression level correlations. To systematically analyze how DEGs with similar expression profiles co-affect OC prognosis, WGCNA (22) was performed. Based on the obtained weighted gene coexpression levels, DEG coexpression was suggested to be significantly associated with OC prognosis. To further identify the genes that were able to differentiate between patients with OC with different prognoses, the verified DEG protein-protein interaction (PPI) pairs were used to construct a PPI network using BioNet 1.24.1 package (http://www.bioconductor.org/packages/release/bioc/html/BioNet.html).

Subnetwork analysis and classifier construction

The BioNet package (http://bionet.bioapps.biozentrum.uni-wuerzburg.de) provides an extensive framework that enables functional subnetworks to be isolated from biological networks. Using the BioNet package in R 3.1.0 (23), as previously described (24), subnetwork analysis was conducted for the PPI network with the P-value/false discovery rate set to 0.01. With the subnetwork nodes as feature genes, a random forest classifier (25) was constructed. For sample labels (recurrence/no recurrence), true and false positive rates were calculated and combined with leave-one-out cross validation (26). Additionally, a receiver operating characteristic (ROC) curve was constructed (27) to evaluate the classification efficiency of the random forest classifier.

Validation using other independent datasets. To confirm that the subnetwork nodes were able to effectively differentiate between patients with OC with different prognoses, the classification efficiency of the random forest classifier for the validation set (GSE44104) was analyzed and presented using a confusion matrix. Additionally, a Kaplan-Meier (KM) survival analysis (28) was performed and combined with the clinical information belonging to the TCGA dataset.

Results

DEG screening

Following implementation of the DEDS algorithm, a total of 44 upregulated and 117 downregulated genes were identified in the recurrent samples relative to the non-recurrent samples, with more downregulated genes identified compared with upregulated genes. Additionally, a volcano plot was constructed to examine DEG expression distributions (Fig. 1).

Functional enrichment analysis

Using the GO Term Finder, significant GO terms were enriched for the upregulated and downregulated genes separately. For the upregulated genes, the enriched GO terms were primarily associated with the ‘regulation of synapse assembly’ (P=1.04×10−7), ‘regulation of synapse organization’ (P=5.26×10−7), and ‘regulation of synapse structure or activity’ (P=5.71×10−7; Table IA). The downregulated genes were associated with ‘single-multicellular organism process’ (P=4.11×10−5), ‘multicellular organismal process’ (P=5.14×10−5) and ‘positive regulation of steroid hormone biosynthetic process’ (P=3.83×10−3; Table IB), which included cytochrome P450 family 17 subfamily A member 1 (CYP17A1).

Table I.

Significant functional terms enriched for the upregulated and downregulated genes.

Table I.

Significant functional terms enriched for the upregulated and downregulated genes.

A, Upregulated genes

TermCorrected P-valueCountGenes
Regulation of synapse assembly 1.04×10−75DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
regulation of synapse organization 5.26×10−75DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
regulation of synapse structure or activity 5.71×10−75DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
synapse assembly 2.08×10−65DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
positive regulation of synapse assembly 6.78×10−64LINGO2, TPBG, SLITRK5, SLITRK3
synapse organization 2.23×10−55DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
regulation of nervous system development 2.72×10−46LRRC7, DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
positive regulation of nervous system development 4.44×10−45LRRC7, LINGO2, TPBG, SLITRK5, SLITRK3
regulation of developmental process 1.131×10−38LRRC7, DKK1, SLITRK3, BVES, LINGO2, MEGF10, SLITRK5, TPBG
regulation of multicellular organismal development 1.96×10−37LRRC7, DKK1, LINGO2, MEGF10, TPBG, SLITRK5, SLITRK3
positive regulation of developmental process 2.64×10−36LRRC7, DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
system development 2.84×10−310LRRC7, DKK1, SLITRK3, WDR69, BVES, C8orf85, LINGO2, MEGF10, TPBG, SLITRK5
regulation of multicellular organismal process 3.63×10−38LRRC7, DKK1, SLITRK3, BVES, LINGO2, MEGF10, SLITRK5, TPBG
single-organism developmental process 5.75×10−311LRRC7, DKK1, SLITRK3, WDR69, BVES, C8orf85, LINGO2, MEGF10, MMP7, TPBG, SLITRK5
developmental process 6.65×10−311LRRC7, DKK1, SLITRK3, WDR69, BVES, C8orf85, LINGO2, MEGF10, MMP7, TPBG, SLITRK5
single-multicellular organism process 8.10×10−311LRRC7, DKK1, SLITRK3, WDR69, BVES, C8orf85, LINGO2, MEGF10, MMP7, TPBG, SLITRK5
positive regulation of multicellular organismal process 8.16×10−36LRRC7, DKK1, LINGO2, TPBG, SLITRK5, SLITRK3
multicellular organism development 9.65×10−310LRRC7, DKK1, SLITRK3, WDR69, BVES, C8orf85, LINGO2, MEGF10, TPBG, SLITRK5
regulation of cellular component biogenesis 9.92×10−35DKK1, LINGO2, TPBG, SLITRK5, SLITRK3

B, Downregulated genes

TermsCorrected P-valueCountGenes

single-multicellular organism process 4.11×10−523GJA8, TRIM71, TCF23, FOXL2, SCNN1G, CPNE5, ARX, CACNA2D2, SLC32A1, AQP5, CCR4, SPTB, HOXC5, RD3, FGF9, PDZD7, PROK1, IL33, WNT6, CYP17A1, BMP6, LGI1, COLEC11
multicellular organismal process 5.14×10−525GJA8, TRIM71, TCF23, FOXL2, SCNN1G, CPNE5, ARX, CACNA2D2, SLC32A1, AQP5, CCR4, TAS1R3, SPTB, HOXC5, RD3, FGF9, PDZD7, PROK1, IL33, WNT6, ACCN3, CYP17A1, BMP6, LGI1, COLEC11
eye development 5.32×10−57RD3, GJA8, FGF9, BMP6, AQP5, FOXL2, WNT6
sensory organ development 7.88×10−58RD3, GJA8, FGF9, BMP6, PDZD7, AQP5, FOXL2, WNT6
multicellular organism development 1.86×10−420GJA8, TRIM71, TCF23, FOXL2, CPNE5, ARX, SLC32A1, AQP5, CCR4, SPTB, HOXC5, FGF9, RD3, PDZD7, PROK1, WNT6, CYP17A1, BMP6, LGI1, COLEC11
anatomical structure development 1.87×10−421GJA8, TRIM71, TCF23, FOXL2, CPNE5, ARX, CACNA2D2, SLC32A1, AQP5, CCR4, SPTB, HOXC5, FGF9, RD3, PDZD7, PROK1, WNT6, CYP17A1, BMP6, LGI1, COLEC11
single-organism developmental process 5.26×10−421GJA8, TRIM71, TCF23, FOXL2, CPNE5, ARX, CACNA2D2, SLC32A1, AQP5, CCR4, SPTB, HOXC5, FGF9, RD3, PDZD7, PROK1, WNT6, CYP17A1, BMP6, LGI1, COLEC11
system development 5.74×10−418GJA8, TRIM71, TCF23, FOXL2, CPNE5, ARX, SLC32A1, AQP5, CCR4, SPTB, FGF9, RD3, PDZD7, PROK1, WNT6, CYP17A1, BMP6, LGI1
developmental process 6.76×10−421GJA8, TRIM71, TCF23, FOXL2, CPNE5, ARX, CACNA2D2, SLC32A1, AQP5, CCR4, SPTB, HOXC5, FGF9, RD3, PDZD7, PROK1, WNT6, CYP17A1, BMP6, LGI1, COLEC11
positive regulation of steroid hormone biosynthetic process 3.83×10−32BMP6, CYP17A1
positive regulation of growth 4.05×10−35CACNA2D2, FGF9, LGI1, CPNE5, ARX
nervous system development 4.48×10−312FGF9, PDZD7, TRIM71, WNT6, CPNE5, CYP17A1, ARX, BMP6, LGI1, SLC32A1, CCR4, SPTB
positive regulation of organ growth 7.93×10−33CACNA2D2, FGF9, ARX
camera-type eye development 8.92×10−35RD3, GJA8, AQP5, FOXL2, WNT6
anatomical structure morphogenesis 9.15×10−312FGF9, PROK1, PDZD7, TRIM71, FOXL2, WNT6, CPNE5, ARX, BMP6, LGI1, AQP5, SPTB
WGCNA

WGCNA was performed and turquoise and grey modules were identified within the cluster dendrogram (Fig. 2). Within the turquoise module, 87 genes were identified (14 upregulated and 73 downregulated), while in the grey module, 74 were identified (30 upregulated and 44 downregulated). Correlations between the two modules regarding status/prognosis were also analyzed. The results demonstrated that the two modules had significant correlations with status and prognosis (P<0.05; Fig. 3). Furthermore, sample correlations were further examined via heatmap analysis, and it was indicated that the samples in the turquoise module were more strongly correlated when compared with those in the grey module (Fig. 4).

Subnetwork analysis and classifier construction

Following building of the PPI network, subnetwork analysis was performed and a significant subnetwork was identified (Fig. 5). The importance scores for the 16 subnetwork nodes [transcription factor GATA-4 (GATA4); fibroblast growth factor 9 (FGF9); aromatase (CYP19A1); 3β-hydroxysteroid dehydrogenase/δ5-4-isomerase type 2 (HSD3B2); corticosteroid 11β-dehydrogenase isozyme 1 (HSD11B1); CYP17A1; pituitary homeobox 2 (PITX2); left-right determination factor 1 (LEFTY1); homeobox protein ARX (ARX); estrogen receptor β (ESR2); steroidogenic factor 1 (NR5A1); forkhead box protein L2 (FOXL2); myocardin (MYOCD); steroidogenic acute regulatory protein mitochondrial (STAR); vesicular inhibitory amino acid transporter (SLC32A1); and twist-related protein 1 (TWIST1)] are listed in Table II. There were multiple interactions among these subnetwork nodes, including HSD3B2-NR5A1, HSD11B1-HSD3B2, CYP17A1-GATA4, ARX-FOXL2, MYOCD-GATA4, STAR-FGF9 and SLC32A1-PITX2. The subnetwork nodes were taken as feature genes and a random forest classifier was constructed. The generated ROC curve demonstrated that the true and false positive rates separately were 92 and 23% when classifying the recurrent and non-recurrent OC samples (Fig. 6A).

Table II.

Importance scores of the subnetwork nodes.

Table II.

Importance scores of the subnetwork nodes.

NodeScore
ARX15.203
CYP17A110.956
CYP19A110.902
ESR210.537
FGF910.148
FOXL29.827
GATA49.786
HSD11B19.223
HSD3B29.196
LEFTY18.259
MYOCD7.867
NR5A17.634
SLC32A16.367
STAR6.167
PITX25.094
TWIST14.708

[i] GATA4, transcription factor GATA-4; FGF9, fibroblast growth factor 9; CYP19A1, aromatase; HSD3B2, 3β-hydroxysteroid dehydrogenase/δ5-4-isomerase type 2; HSD11B1, corticosteroid 11β-dehydrogenase isozyme 1; CYP17A1, cytochrome P450 family 17 subfamily A member 1; PITX2, pituitary homeobox 2; LEFTY1, left-right determination factor 1; ARX, homeobox protein ARX; ESR2, estrogen receptor β; NR5A1, steroidogenic factor 1; FOXL2, forkhead box protein L2; MYOCD, myocardin; STAR, steroidogenic acute regulatory protein mitochondrial; SLC32A1, vesicular inhibitory amino acid transporter; TWIST1, twist-related protein 1.

Validation using other independent datasets

The random forest classifier was used to differentiate between samples in the GSE44104 validation set, and the prediction accuracies for the non-recurrence and recurrence groups were 87.5 and 85%, respectively (Fig. 6B). These findings indicated that the subnetwork nodes were important in predicting OC prognosis.

Of the 307 OC samples in the TCGA dataset, only 262 samples remained upon removal of samples without follow-up or survival time information. These 262 samples were divided into high- and low-risk groups using the random forest classifier, and further examined using KM survival analysis. The results demonstrated that the low risk group had a significantly longer survival time compared with the high-risk group (P=0.0166; Fig. 7A). Subsequently, the samples from the second validation set (GSE49997) were divided into high- and low-risk groups using the random forest classifier. Following analysis using a KM survival curve, a significant difference was noted in survival time between the low- and high-risk groups (P=0.0165; Fig. 7B). These findings suggested that the random forest classifier had portability and repeatability.

Discussion

In the present study, 44 upregulated and 117 downregulated genes were identified in the recurrent samples relative to the non-recurrent samples. When performing WGCNA, turquoise and grey modules were identified that had significant correlations with status and prognosis. Furthermore, a significant subnetwork was identified from the PPI network, with the subnetwork nodes (including GATA4, FGF9, CYP19A1, HSD3B2, HSD11B1, CYP17A1, PITX2, LEFTY1, ARX, ESR2, NR5A1, FOXL2, MYOCD, STAR, SLC32A1 and TWIST1) being utilized as feature genes for constructing a random forest classifier. Moreover, the classification efficiency of the random forest classifier was validated and confirmed.

Of the identified nodes, previous studies have reported that GATA4 overexpression, in conjunction with human epidermal growth factor receptor 2, may predict a shorter disease-free survival time and may be utilized as a prognostic marker in patients with ovarian granulosa cell tumor to optimize follow-up management in the early stages (29,30). In OC, GATA4 and transcription factor GATA-6 expression is frequently lost, with this feature specifying the histological subtype prior to tumorigenic transition of the ovarian surface epithelium (31). The upregulation of FGF9 has been detected in primary ovarian endometrioid adenocarcinomas carrying a defective Wnt/β-catenin pathway and serves an important role in promoting the cancer phenotype (32,33). The aromatase enzyme encoded by the CYP19A1 gene acts in the conversion of androgen to estrogen, with CYP19A1 variants potentially able to influence OC susceptibility (34). Thus, GATA4, FGF9 and CYP19A1 appear to be implicated in the mechanisms of OC pathogenesis. Enrichment analysis demonstrated that CYP17A1, which is involved in ‘positive regulation of steroid hormone biosynthetic processes’, was downregulated. A previous study demonstrated that steroid hormones serve a role in OC pathogenesis, and their receptors influence OC patient survival (35). Therefore, CYP17A1 may affect OC patient prognoses through the positive regulation of steroid hormones.

The overexpression of PITX2 has been implicated in OC progression by facilitating cell growth, migration and invasion, and thus may potentially serve as a therapeutic target for patients with high-grade OC (36,37). PITX2 has also been demonstrated to promote OC cell proliferation through the Wnt pathway, which is closely associated with ovarian development and OC (38). In ovarian clear cell carcinoma, the overexpression of LEFTY, a transforming growth factor-β superfamily member, exhibits an anti-tumor effect by affecting cell proliferation and cellular susceptibility to apoptotic signals (39). Estrogen receptor β, which is encoded by the ESR2 gene, has been suggested to serve as a critical factor during OC carcinogenesis (40). Furthermore, within the ESR2 promoter region, the genotypic and allelic frequencies of the single nucleotide polymorphism (SNP) rs3020449 have been demonstrated to exhibit significant differences based on OC stage, thus indicating that SNP rs3020449 may be associated with OC progression (41). These findings suggest that PITX2, LEFTY1 and ESR2 may also serve roles in OC pathogenesis.

NR5A1 serves an important role in ovarian function and development, with an NR5A1 mutation reported to induce 46 XY disorders of sex development (42). FOXL2 is critical for GC (granulosa cell) differentiation during the process of folliculogenesis, with its downregulation potentially serving as an ovarian granulosa cells tumor prognostic factor (43). TWIST1, which may induce epithelial-mesenchymal transition and contribute to tumor metastasis, has been demonstrated to be associated with poor survival in patients with cancer (44,45). Thus, NR5A1, FOXL2 and TWIST1 may be associated with patient survival in OC. Moreover, the multiple interactions within the PPI subnetwork (including HSD3B2-NR5A1, HSD11B1-HSD3B2, CYP17A1-GATA4, ARX-FOXL2, MYOCD-GATA4, STAR-FGF9 and SLC32A1-PITX2) indicate that HSD3B2, HSD11B1, CYP17A1, ARX, MYOCD, STAR and SLC32A1 may also function in OC by interacting with other genes.

However, the present study has several limitations to note. First, the datasets used in the present study had sample size differences, platform differences and data heterogeneities that may affect the prediction accuracy of the random forest classifier. Second, the smaller patient numbers and analytical methods may limit the predictive capability of the present model. Finally, only bioinformatics analyses were conducted in the present study, and no direct experimental validation was performed. Therefore, further analyses are required to validate the obtained results.

In conclusion, 44 upregulated and 117 downregulated genes associated with OC recurrence were identified. Furthermore, the 16 subnetwork node genes that were identified may be critical molecules associated with OC recurrence.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the Hubei Province's Outstanding Medical Academic Leader Program and the Hubei Province Health and Family Planning Scientific Research Project (grant no. WJ2015MA024) and the Natural Science Foundation of Hubei Province (grant no. 2017CFB335).

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LC and LL performed data analyses and wrote the manuscript. LW, XL and HX contributed significantly in data analyses and manuscript revision. JZ conceived and designed the study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jayson GC, Kohn EC, Kitchener HC and Ledermann JA: Ovarian cancer. Lancet. 384:1376–1388. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Ebell MH, Culp MB and Radke TJ: A systematic review of symptoms for the diagnosis of ovarian cancer. Am J Prev Med. 50:384–394. 2016. View Article : Google Scholar : PubMed/NCBI

3 

McGuire S: World Cancer Report 2014. Geneva, Switzerland: World Health Organization, International Agency for Research on Cancer, WHO Press, 2015. Adv Nutr. 7:418–419. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Lengyel E: ovarian cancer development and metastasis. Am J Pathol. 177:1053–1064. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Cree IA: Cancer biology. Methods Mol Biol. 731:1–11. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Holschneider CH and Berek JS: Ovarian cancer: Epidemiology, biology, and prognostic factors. Semin Surg Oncol. 19:3–10. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Tworoger SS and Doherty JA: Epidemiologic paradigms for progress in ovarian cancer research. Cancer Causes Control. 28:361–364. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Siu MK, Chan HY, Kong DS, Wong ES, Wong OG, Ngan HY, Tam KF, Zhang H, Li Z, Chan QK, et al: p21-activated kinase 4 regulates ovarian cancer cell proliferation, migration, and invasion and contributes to poor prognosis in patients. Proc Natl Acad Sci USA. 107:18622–18627. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Nakayama N, Nakayama K, Shamima Y, Ishikawa M, Katagiri A, Iida K and Miyazaki K: Gene amplification CCNE1 is related to poor survival and potential therapeutic target in ovarian cancer. Cancer. 116:2621–2634. 2010.PubMed/NCBI

10 

Etemadmoghadam D, George J, Cowin PA, Cullinane C and Kansara M: Australian Ovarian Cancer Study Group, Gorringe KL, Smyth GK and Bowtell DD: Amplicon-dependent CCNE1 expression is critical for clonogenic survival after cisplatin treatment and is correlated with 20q11 gain in ovarian cancer. PLoS One. 5:e154982010. View Article : Google Scholar : PubMed/NCBI

11 

Ehlén Å, Brennan DJ, Nodin B, O'Connor DP, Eberhard J, Alvarado-Kristensson M, Jeffrey IB, Manjer J, Brändstedt J, Uhlén M, et al: Expression of the RNA-binding protein RBM3 is associated with a favourable prognosis and cisplatin sensitivity in epithelial ovarian cancer. J Transl Med. 8:782010. View Article : Google Scholar : PubMed/NCBI

12 

Xia Y, Chang T, Wang Y, Liu Y, Li W, Li M and Fan HY: YAP promotes ovarian cancer cell tumorigenesis and is indicative of a poor prognosis for ovarian cancer patients. PLoS One. 9:e917702014. View Article : Google Scholar : PubMed/NCBI

13 

Zhang X, George J, Deb S, Degoutin JL, Takano EA and Fox SB: AOCS Study group, Bowtell DD and Harvey KF: The Hippo pathway transcriptional co-activator, YAP, is an ovarian cancer oncogene. Oncogene. 30:2810–2822. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Zhang J, Guo X, Chang DY, Rosen DG, Mercado-Uribe I and Liu J: CD133 expression associated with poor prognosis in ovarian cancer. Mod Pathol. 25:456–464. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Servant N, Roméjon J, Gestraud P, La Rosa P, Lucotte G, Lair S, Bernard V, Zeitouni B, Coffin F, Jules-Clément G, et al: Bioinformatics for precision medicine in oncology: Principles and application to the SHIVA clinical trial. Front Genet. 5:1522014. View Article : Google Scholar : PubMed/NCBI

16 

Altshuler D, Daly MJ and Lander ES: Genetic mapping in human disease. Science. 322:881–888. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Chen JM, Cooper DN, Chuzhanova N, Férec C and Patrinos GP: Gene conversion: Mechanisms, evolution and human disease. Nat Rev Genet. 8:762–775. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Segundo ES, Tsanas A and Gómez-Vilda P: Euclidean distances as measures of speaker dissimilarity including identical twin pairs: A forensic investigation using source and filter voice characteristics. Forensic Sci Int. 270:25–38. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Yang YH, Xiao Y and Segal MR: Identifying differentially expressed genes from microarray experiments via statistic synthesis. Bioinformatics. 21:1084–1093. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Tweedie S, Ashburner M, Falls K, Leyland P, McQuilton P, Marygold S, Millburn G, Osumi-Sutherland D, Schroeder A, Seal R, et al: FlyBase: Enhancing Drosophila gene ontology annotations. Nucleic Acids Res. 37:(Database issue). D555–D559. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Lässer C, O'Neil SE, Shelke GV, Sihlbom C, Hansson SF, Gho YS, Lundbäck B and Lötvall J: Exosomes in the nose induce immune cell trafficking and harbour an altered protein cargo in chronic airway inflammation. J Transl Med. 14:1812016. View Article : Google Scholar : PubMed/NCBI

22 

Iancu OD, Colville A, Oberbeck D, Darakjian P, Mcweeney SK and Hitzemann R: Cosplicing network analysis of mammalian brain RNA-Seq data utilizing WGCNA and Mantel correlations. Front Genet. 6:1742015. View Article : Google Scholar : PubMed/NCBI

23 

RC Team: R: A language and environment for statistical computingR Foundation for Statistical Computing. Vienna, Austria: 2016

24 

Beisser D, Klau GW, Dandekar T, Müller T and Dittrich MT: BioNet: An R-Package for the functional analysis of biological networks. Bioinformatics. 26:1129–1130. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Ma X, Guo J and Sun X: DNABP: Identification of DNA-binding proteins based on feature selection using a random forest and predicting binding residues. PLoS One. 11:e01673452016. View Article : Google Scholar : PubMed/NCBI

26 

Mikshowsky A, Weigel KA and Gianola D: 0294 Assessing genomic prediction accuracy for Holstein sires using bootstrap aggregation sampling and leave-one-out cross validation. J Anim Sci. 94:139–140. 2016. View Article : Google Scholar

27 

Thomas M: ROC curve comparison of ICNARC and POSSUM in a UK critical care unit. Br J Anaesth. 117:6672016. View Article : Google Scholar : PubMed/NCBI

28 

May WL: Kaplan-Meier survival analysisEncyclopedia of Cancer. Schwab M: Springer; Berlin: pp. 1934–1937. 2011, View Article : Google Scholar

29 

Färkkilä A, Andersson N, Bützow R, Leminen A, Heikinheimo M, Anttonen M and Unkila-Kallio L: HER2 and GATA4 are new prognostic factors for early-stage ovarian granulosa cell tumor-a long-term follow-up study. Cancer Med. 3:526–536. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Anttonen M, Pihlajoki M, Andersson N, Georges A, L'hôte D, Vattulainen S, Färkkilä A1, Unkila-Kallio L, Veitia RA and Heikinheimo M: FOXL2, GATA4, and SMAD3 co-operatively modulate gene expression, cell viability and apoptosis in ovarian granulosa cell tumor cells. PLoS One. 9:e855452014. View Article : Google Scholar : PubMed/NCBI

31 

Cai KQ, Caslini C, Capo-Chichi CD, Slater C, Smith ER, Wu H, Klein-Szanto AJ, Godwin AK and Xu XX: Loss of GATA4 and GATA6 expression specifies ovarian cancer histological subtypes and precedes neoplastic transformation of ovarian surface epithelia. PLoS One. 4:e64542009. View Article : Google Scholar : PubMed/NCBI

32 

Hendrix ND, Wu R, Kuick R, Schwartz DR, Fearon ER and Cho KR: Fibroblast growth factor 9 has oncogenic activity and is a downstream target of Wnt signaling in ovarian endometrioid adenocarcinomas. Cancer Res. 66:1354–1362. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Drummond AE, Tellbach M, Dyson M and Findlay JK: Fibroblast growth factor-9, a local regulator of ovarian function. Endocrinology. 148:3711–3721. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Goodman MT, Lurie G, Thompson PJ, Mcduffie KE and Carney ME: Association of two common single-nucleotide polymorphisms in the CYP19A1 locus and ovarian cancer risk. Endocr Relat Cancer. 15:1055–1060. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Jönsson JM, Arildsen NS, Malander S, Måsbäck A, Hartman L, Nilbert M and Hedenfalk I: Sex steroid hormone receptor expression affects ovarian cancer survival. Transl Oncol. 8:424–433. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Fung FK, Chan DW, Liu VW, Leung TH, Cheung AN and Ngan HY: Increased expression of PITX2 transcription factor contributes to ovarian cancer progression. PLoS One. 7:e370762012. View Article : Google Scholar : PubMed/NCBI

37 

Basu M, Bhattacharya R, Ray U, Mukhopadhyay S, Chatterjee U and Roy SS: Invasion of ovarian cancer cells is induced byPITX2-mediated activation of TGF-β and Activin-A. Mol Cancer. 14:1622015. View Article : Google Scholar : PubMed/NCBI

38 

Basu M and Roy SS: Wnt/β-catenin pathway is regulated by PITX2 homeodomain protein and thus contributes to the proliferation of human ovarian adenocarcinoma cell, SKOV-3. J Biol Chem. 288:4355–4367. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Akiya M, Yamazaki M, Matsumoto T, Kawashima Y, Oguri Y, Kajita S, Kijima D, Chiba R, Yokoi A, Takahashi H, et al: Identification of LEFTY as a molecular marker for ovarian clear cell carcinoma. Oncotarget. 8:63646–63664. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Pearce Leigh C, Near AM, Butler JL, Van Den Berg D, Bretsky P, Conti DV, Stram DO, Pike MC, Hirschhorn JN and Wu AH: Comprehensive evaluation of ESR2 variation and ovarian cancer risk. Cancer Epidemiol Biomarkers Prev. 17:393–396. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Schüler S, Lattrich C, Skrzypczak M, Fehm T, Ortmann O and Treeck O: Polymorphisms in the promoter region of ESR2 gene and susceptibility to ovarian cancer. Gene. 546:283–287. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Lourenço D, Brauner R, Lin L, De Perdigo A, Weryha G, Muresan M, Boudjenah R, Guerra-Junior G, Maciel-Guerra AT, Achermann JC, et al: Mutations in NR5A1 Associated with Ovarian Insufficiency. N Engl J Med. 360:1200–1210. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Kalfa N, Philibert P, Patte C, Ecochard A, Duvillard P, Baldet P, Jaubert F, Fellous M and Sultan C: Extinction of FOXL2 expression in aggressive ovarian granulosa cell tumors in children. Fertil Steril. 87:896–901. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Wushou A, Hou J, Zhao YJ and Shao ZM: Twist-1 up-regulation in carcinoma correlates to poor survival. Int J Mol Sci. 15:21621–21630. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Yin G, Alvero AB, Craveiro V, Holmberg JC, Fu HH, Montagna MK, Yang Y, Chefetz-Menaker I, Nuti S, Rossi M, et al: Constitutive proteasomal degradation of TWIST-1 in epithelial-ovarian cancer stem cells impacts differentiation and metastatic potential. Oncogene. 32:39–49. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 18 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cheng L, Li L, Wang L, Li X, Xing H and Zhou J: A random forest classifier predicts recurrence risk in patients with ovarian cancer. Mol Med Rep 18: 3289-3297, 2018
APA
Cheng, L., Li, L., Wang, L., Li, X., Xing, H., & Zhou, J. (2018). A random forest classifier predicts recurrence risk in patients with ovarian cancer. Molecular Medicine Reports, 18, 3289-3297. https://doi.org/10.3892/mmr.2018.9300
MLA
Cheng, L., Li, L., Wang, L., Li, X., Xing, H., Zhou, J."A random forest classifier predicts recurrence risk in patients with ovarian cancer". Molecular Medicine Reports 18.3 (2018): 3289-3297.
Chicago
Cheng, L., Li, L., Wang, L., Li, X., Xing, H., Zhou, J."A random forest classifier predicts recurrence risk in patients with ovarian cancer". Molecular Medicine Reports 18, no. 3 (2018): 3289-3297. https://doi.org/10.3892/mmr.2018.9300